Protein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells

In the absence of pregnancy the ovarian corpus luteum undergoes regression, a process characterized by decreased production of progesterone and structural luteolysis involving apoptosis. Autophagy has been observed in the corpus luteum during luteal regression. Autophagy is a self-degradative proces...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Emilia Przygrodzka, Corrine F. Monaco, Michele R. Plewes, Guojuan Li, Jennifer R. Wood, Andrea S. Cupp, John S. Davis
Formato: article
Lenguaje:EN
Publicado: Frontiers Media S.A. 2021
Materias:
PKA
Acceso en línea:https://doaj.org/article/28e2fbd226b64c4d9ded9f08b88a83ea
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:28e2fbd226b64c4d9ded9f08b88a83ea
record_format dspace
spelling oai:doaj.org-article:28e2fbd226b64c4d9ded9f08b88a83ea2021-11-08T13:47:26ZProtein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells2296-634X10.3389/fcell.2021.723563https://doaj.org/article/28e2fbd226b64c4d9ded9f08b88a83ea2021-11-01T00:00:00Zhttps://www.frontiersin.org/articles/10.3389/fcell.2021.723563/fullhttps://doaj.org/toc/2296-634XIn the absence of pregnancy the ovarian corpus luteum undergoes regression, a process characterized by decreased production of progesterone and structural luteolysis involving apoptosis. Autophagy has been observed in the corpus luteum during luteal regression. Autophagy is a self-degradative process important for balancing sources of cellular energy at critical times in development and in response to nutrient stress, but it can also lead to apoptosis. Mechanistic target of rapamycin (MTOR) and 5′ AMP-activated protein kinase (AMPK), key players in autophagy, are known to inhibit or activate autophagy, respectively. Here, we analyzed the signaling pathways regulating the initiation of autophagy in bovine luteal cells. In vivo studies showed increased activating phosphorylation of AMPKα (Thr172) and elevated content of LC3B, a known marker of autophagy, in luteal tissue during PGF2α-induced luteolysis. In vitro, AMPK activators 1) stimulated phosphorylation of regulatory associated protein of MTOR (RPTOR) leading to decreased activity of MTOR, 2) increased phosphorylation of Unc-51-Like Kinase 1 (ULK1) and Beclin 1 (BECN1), at sites specific for AMPK and required for autophagy initiation, 3) increased levels of LC3B, and 4) enhanced colocalization of autophagosomes with lysosomes indicating elevated autophagy. In contrast, LH/PKA signaling in luteal cells 1) reduced activation of AMPKα and phosphorylation of RPTOR, 2) elevated MTOR activity, 3) stimulated phosphorylation of ULK1 at site required for ULK1 inactivation, and 4) inhibited autophagosome formation as reflected by reduced content of LC3B-II. Pretreatment with AICAR, a pharmacological activator of AMPK, inhibited LH-mediated effects on RPTOR, ULK1 and BECN1. Our results indicate that luteotrophic signaling via LH/PKA/MTOR inhibits, while luteolytic signaling via PGF2α/Ca2+/AMPK activates key signaling pathways involved in luteal cell autophagy.Emilia PrzygrodzkaCorrine F. MonacoCorrine F. MonacoMichele R. PlewesMichele R. PlewesGuojuan LiJennifer R. WoodAndrea S. CuppAndrea S. CuppJohn S. DavisJohn S. DavisFrontiers Media S.A.articlecorpus luteumPGF2αluteinizing hormoneAMPKPKAMTORBiology (General)QH301-705.5ENFrontiers in Cell and Developmental Biology, Vol 9 (2021)
institution DOAJ
collection DOAJ
language EN
topic corpus luteum
PGF2α
luteinizing hormone
AMPK
PKA
MTOR
Biology (General)
QH301-705.5
spellingShingle corpus luteum
PGF2α
luteinizing hormone
AMPK
PKA
MTOR
Biology (General)
QH301-705.5
Emilia Przygrodzka
Corrine F. Monaco
Corrine F. Monaco
Michele R. Plewes
Michele R. Plewes
Guojuan Li
Jennifer R. Wood
Andrea S. Cupp
Andrea S. Cupp
John S. Davis
John S. Davis
Protein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells
description In the absence of pregnancy the ovarian corpus luteum undergoes regression, a process characterized by decreased production of progesterone and structural luteolysis involving apoptosis. Autophagy has been observed in the corpus luteum during luteal regression. Autophagy is a self-degradative process important for balancing sources of cellular energy at critical times in development and in response to nutrient stress, but it can also lead to apoptosis. Mechanistic target of rapamycin (MTOR) and 5′ AMP-activated protein kinase (AMPK), key players in autophagy, are known to inhibit or activate autophagy, respectively. Here, we analyzed the signaling pathways regulating the initiation of autophagy in bovine luteal cells. In vivo studies showed increased activating phosphorylation of AMPKα (Thr172) and elevated content of LC3B, a known marker of autophagy, in luteal tissue during PGF2α-induced luteolysis. In vitro, AMPK activators 1) stimulated phosphorylation of regulatory associated protein of MTOR (RPTOR) leading to decreased activity of MTOR, 2) increased phosphorylation of Unc-51-Like Kinase 1 (ULK1) and Beclin 1 (BECN1), at sites specific for AMPK and required for autophagy initiation, 3) increased levels of LC3B, and 4) enhanced colocalization of autophagosomes with lysosomes indicating elevated autophagy. In contrast, LH/PKA signaling in luteal cells 1) reduced activation of AMPKα and phosphorylation of RPTOR, 2) elevated MTOR activity, 3) stimulated phosphorylation of ULK1 at site required for ULK1 inactivation, and 4) inhibited autophagosome formation as reflected by reduced content of LC3B-II. Pretreatment with AICAR, a pharmacological activator of AMPK, inhibited LH-mediated effects on RPTOR, ULK1 and BECN1. Our results indicate that luteotrophic signaling via LH/PKA/MTOR inhibits, while luteolytic signaling via PGF2α/Ca2+/AMPK activates key signaling pathways involved in luteal cell autophagy.
format article
author Emilia Przygrodzka
Corrine F. Monaco
Corrine F. Monaco
Michele R. Plewes
Michele R. Plewes
Guojuan Li
Jennifer R. Wood
Andrea S. Cupp
Andrea S. Cupp
John S. Davis
John S. Davis
author_facet Emilia Przygrodzka
Corrine F. Monaco
Corrine F. Monaco
Michele R. Plewes
Michele R. Plewes
Guojuan Li
Jennifer R. Wood
Andrea S. Cupp
Andrea S. Cupp
John S. Davis
John S. Davis
author_sort Emilia Przygrodzka
title Protein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells
title_short Protein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells
title_full Protein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells
title_fullStr Protein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells
title_full_unstemmed Protein Kinase A and 5′ AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells
title_sort protein kinase a and 5′ amp-activated protein kinase signaling pathways exert opposite effects on induction of autophagy in luteal cells
publisher Frontiers Media S.A.
publishDate 2021
url https://doaj.org/article/28e2fbd226b64c4d9ded9f08b88a83ea
work_keys_str_mv AT emiliaprzygrodzka proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT corrinefmonaco proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT corrinefmonaco proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT michelerplewes proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT michelerplewes proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT guojuanli proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT jenniferrwood proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT andreascupp proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT andreascupp proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT johnsdavis proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
AT johnsdavis proteinkinaseaand5ampactivatedproteinkinasesignalingpathwaysexertoppositeeffectsoninductionofautophagyinlutealcells
_version_ 1718442217917382656